Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
PLoS Genet ; 20(4): e1011235, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38648200

RESUMEN

Tumor-associated macrophages (TAM) subtypes have been shown to impact cancer prognosis and resistance to immunotherapy. However, there is still a lack of systematic investigation into their molecular characteristics and clinical relevance in different cancer types. Single-cell RNA sequencing data from three different tumor types were used to cluster and type macrophages. Functional analysis and communication of TAM subpopulations were performed by Gene Ontology-Biological Process and CellChat respectively. Differential expression of characteristic genes in subpopulations was calculated using zscore as well as edgeR and Wilcoxon rank sum tests, and subsequently gene enrichment analysis of characteristic genes and anti-PD-1 resistance was performed by the REACTOME database. We revealed the heterogeneity of TAM, and identified eleven subtypes and their impact on prognosis. These subtypes expressed different molecular functions respectively, such as being involved in T cell activation, apoptosis and differentiation, or regulating viral bioprocesses or responses to viruses. The SPP1 pathway was identified as a critical mediator of communication between TAM subpopulations, as well as between TAM and epithelial cells. Macrophages with high expression of SPP1 resulted in poorer survival. By in vitro study, we showed SPP1 mediated the interactions between TAM clusters and between TAM and tumor cells. SPP1 promoted the tumor-promoting ability of TAM, and increased PDL1 expression and stemness of tumor cells. Inhibition of SPP1 attenuated N-cadherin and ß-catenin expression and the activation of AKT and STAT3 pathway in tumor cells. Additionally, we found that several subpopulations could decrease the sensitivity of anti-PD-1 therapy in melanoma. SPP1 signal was a critical pathway of communication between macrophage subtypes. Some specific macrophage subtypes were associated with immunotherapy resistance and prognosis in some cancer types.


Asunto(s)
Neoplasias , Osteopontina , Macrófagos Asociados a Tumores , Humanos , Macrófagos Asociados a Tumores/inmunología , Macrófagos Asociados a Tumores/metabolismo , Pronóstico , Neoplasias/inmunología , Neoplasias/genética , Osteopontina/genética , Osteopontina/metabolismo , Regulación Neoplásica de la Expresión Génica , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Línea Celular Tumoral , beta Catenina/genética , beta Catenina/metabolismo , Análisis de la Célula Individual , Transducción de Señal , Macrófagos/inmunología , Macrófagos/metabolismo , Comunicación Celular/inmunología
2.
Apoptosis ; 29(9-10): 1499-1514, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-38853202

RESUMEN

Ovarian cancer is a malignant tumor originating from the ovary, characterized by its high mortality rate and propensity for recurrence. In some patients, especially those with recurrent cancer, conventional treatments such as surgical resection or standard chemotherapy yield suboptimal results. Consequently, there is an urgent need for novel anti-cancer therapeutic strategies. Ferroptosis is a distinct form of cell death separate from apoptosis. Ferroptosis inducers have demonstrated promising potential in the treatment of ovarian cancer, with evidence indicating their ability to enhance ovarian cancer cell sensitivity to cisplatin. However, resistance of cancer cells to ferroptosis still remains an inevitable challenge. Here, we analyzed genome-scale CRISPR-Cas9 loss-of function screens and identified PAX8 as a ferroptosis resistance protein in ovarian cancer. We identified PAX8 as a susceptibility gene in GPX4-dependent ovarian cancer. Depletion of PAX8 rendered GPX4-dependent ovarian cancer cells significantly more sensitive to GPX4 inhibitors. Additionally, we found that PAX8 inhibited ferroptosis in ovarian cancer cells. Combined treatment with a PAX8 inhibitor and RSL3 suppressed ovarian cancer cell growth, induced ferroptosis, and was validated in a xenograft mouse model. Further exploration of the molecular mechanisms underlying PAX8 inhibition of ferroptosis mutations revealed upregulation of glutamate-cysteine ligase catalytic subunit (GCLC) expression. GCLC mediated the ferroptosis resistance induced by PAX8 in ovarian cancer. In conclusion, our study underscores the pivotal role of PAX8 as a therapeutic target in GPX4-dependent ovarian cancer. The combination of PAX8 inhibitors such as losartan and captopril with ferroptosis inducers represents a promising new approach for ovarian cancer therapy.


Asunto(s)
Ferroptosis , Glutatión , Neoplasias Ováricas , Factor de Transcripción PAX8 , Fosfolípido Hidroperóxido Glutatión Peroxidasa , Ensayos Antitumor por Modelo de Xenoinjerto , Ferroptosis/efectos de los fármacos , Ferroptosis/genética , Humanos , Femenino , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Factor de Transcripción PAX8/genética , Factor de Transcripción PAX8/metabolismo , Animales , Línea Celular Tumoral , Ratones , Glutatión/metabolismo , Fosfolípido Hidroperóxido Glutatión Peroxidasa/metabolismo , Fosfolípido Hidroperóxido Glutatión Peroxidasa/genética , Ratones Desnudos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Sistemas CRISPR-Cas , Proliferación Celular/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Carbolinas
3.
Apoptosis ; 29(5-6): 663-680, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38598070

RESUMEN

Cancer cachexia-associated muscle wasting as a multifactorial wasting syndrome, is an important factor affecting the long-term survival rate of tumor patients. Photobiomodulation therapy (PBMT) has emerged as a promising tool to cure and prevent many diseases. However, the effect of PBMT on skeletal muscle atrophy during cancer progression has not been fully demonstrated yet. Here, we found PBMT alleviated the atrophy of myotube diameter induced by cancer cells in vitro, and prevented cancer-associated muscle atrophy in mice bearing tumor. Mechanistically, the alleviation of muscle wasting by PBMT was found to be involved in inhibiting E3 ubiquitin ligases MAFbx and MuRF-1. In addition, transcriptomic analysis using RNA-seq and GSEA revealed that PI3K/AKT pathway might be involved in PBMT-prevented muscle cachexia. Next, we showed the protective effect of PBMT against muscle cachexia was totally blocked by AKT inhibitor in vitro and in vivo. Moreover, PBMT-activated AKT promoted FoxO3a phosphorylation and thus inhibiting the nucleus entry of FoxO3a. Lastly, in cisplatin-treated muscle cachexia model, PBMT had also been shown to ameliorate muscle atrophy through enhancing PI3K/AKT pathway to suppress MAFbx and MuRF-1 expression. These novel findings revealed that PBMT could be a promising therapeutic approach in treating muscle cachexia induced by cancer.


Asunto(s)
Caquexia , Proteína Forkhead Box O3 , Enfermedades Musculares , Neoplasias , Fosfatidilinositol 3-Quinasas , Proteínas Proto-Oncogénicas c-akt , Síndrome Debilitante , Caquexia/etiología , Caquexia/metabolismo , Caquexia/terapia , Enfermedades Musculares/etiología , Enfermedades Musculares/metabolismo , Enfermedades Musculares/terapia , Neoplasias/complicaciones , Redes y Vías Metabólicas , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteína Forkhead Box O3/genética , Proteína Forkhead Box O3/metabolismo , Síndrome Debilitante/etiología , Síndrome Debilitante/metabolismo , Síndrome Debilitante/terapia , Animales , Modelos Animales de Enfermedad , Ratones , Línea Celular , Masculino , Ratones Endogámicos BALB C , Perfilación de la Expresión Génica
4.
Apoptosis ; 28(1-2): 81-107, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36399287

RESUMEN

It has been 10 years since the concept of ferroptosis was put forward and research focusing on ferroptosis has been increasing continuously. Ferroptosis is driven by iron-dependent lipid peroxidation, which can be antagonized by glutathione peroxidase 4 (GPX4), ferroptosis inhibitory protein 1 (FSP1), dihydroorotate dehydrogenase (DHODH) and Fas-associated factor 1 (FAF1). Various cellular metabolic events, including lipid metabolism, can modulate ferroptosis sensitivity. It is worth noting that the reprogramming of lipid metabolism in cancer cells can promote the occurrence and development of tumors. The metabolic flexibility of cancer cells opens the possibility for the coordinated targeting of multiple lipid metabolic pathways to trigger cancer cells ferroptosis. In addition, cancer cells must obtain immortality, escape from programmed cell death including ferroptosis, to promote cancer progression, which provides new perspectives for improving cancer therapy. Targeting the vulnerability of ferroptosis has received attention as one of the significant possible strategies to treat cancer given its role in regulating tumor cell survival. We review the impact of iron and lipid metabolism on ferroptosis and the potential role of the crosstalk of lipid metabolism reprogramming and ferroptosis in antitumor immunity and sum up agents targeting lipid metabolism and ferroptosis for cancer therapy.


Asunto(s)
Ferroptosis , Neoplasias , Humanos , Apoptosis , Metabolismo de los Lípidos , Peroxidación de Lípido , Neoplasias/metabolismo , Hierro/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Reguladoras de la Apoptosis/metabolismo
5.
J Biomed Sci ; 30(1): 8, 2023 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-36707854

RESUMEN

Chronic stress results in disturbances of body hormones through the neuroendocrine system. Cancer patients often experience recurrent anxiety and restlessness during disease progression and treatment, which aggravates disease progression and hinders treatment effects. Recent studies have shown that chronic stress-regulated neuroendocrine systems secret hormones to activate many signaling pathways related to tumor development in tumor cells. The activated neuroendocrine system acts not only on tumor cells but also modulates the survival and metabolic changes of surrounding non-cancerous cells. Current clinical evidences also suggest that chronic stress affects the outcome of cancer treatment. However, in clinic, there is lack of effective treatment for chronic stress in cancer patients. In this review, we discuss the main mechanisms by which chronic stress regulates the tumor microenvironment, including functional regulation of tumor cells by stress hormones (stem cell-like properties, metastasis, angiogenesis, DNA damage accumulation, and apoptotic resistance), metabolic reprogramming and immune escape, and peritumor neuromodulation. Based on the current clinical treatment framework for cancer and chronic stress, we also summarize pharmacological and non-pharmacological therapeutic approaches to provide some directions for cancer therapy.


Asunto(s)
Neoplasias , Humanos , Neoplasias/metabolismo , Transducción de Señal , Progresión de la Enfermedad , Hormonas/farmacología , Microambiente Tumoral
6.
Apoptosis ; 27(11-12): 946-960, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36028785

RESUMEN

Developing individualized therapies for different renal cell carcinoma patients is pivotal for improving the efficacy of immunotherapy. It has been reported that ferroptosis is involved in T cell-mediated anti-tumor immunity, and that therapeutic approaches targeting tumor ferroptosis pathway in combination with immune checkpoint blockade drugs improve the efficacy of cancer immunotherapy. This study focused specifically on ferroptosis genes to identify novel biomarkers that reflect prognosis in different renal cell carcinoma subtypes. LASSO algorithm and multivariate Cox regression were initiated for identifying ferroptosis-related multigene risk signature (FRGsig) and established a FRGsig score model. We used multiple tumor microenvironment gene signatures and methods to infer tumor microenvironment status and immune cell invasion levels. Our study found that high FRGsig score was associated with poor prognosis in patients with predominant histologic subtypes of renal cell carcinoma. And high FRGsig score samples had higher levels of anti-tumor immunity cells infiltration, and there was a feedback mechanism whereby anti-tumor inflammation promoted the recruitment or differentiation of immunosuppressive cells. FRGsig was a potential biomarker for predicting the response to immune checkpoint blockade therapy in kidney clear cell carcinoma and kidney papillary cell carcinoma, and the kidney papillary cell carcinoma patients with high FRGsig was associated with better response to anti-VEGF therapy. Our findings provided further insights into assessing immunotherapy sensitivity of predominant histologic subtypes of renal cell carcinoma. FRGsig might be a potential biomarker for predicting the efficacy of angiogenic blocking drugs or immune checkpoint inhibitors in different renal cell carcinoma subtypes, enabling more precise patient selection.


Asunto(s)
Carcinoma de Células Renales , Ferroptosis , Neoplasias Renales , Humanos , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/terapia , Ferroptosis/genética , Inhibidores de Puntos de Control Inmunológico , Apoptosis , Inmunoterapia , Microambiente Tumoral/genética , Neoplasias Renales/genética , Neoplasias Renales/terapia
7.
Biochem J ; 478(9): 1663-1688, 2021 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-33970218

RESUMEN

Cancer cachexia often occurs in malignant tumors and is a multifactorial and complex symptom characterized by wasting of skeletal muscle and adipose tissue, resulting in weight loss, poor life quality and shorter survival. The pathogenic mechanism of cancer cachexia is complex, involving a variety of molecular substrates and signal pathways. Advancements in understanding the molecular mechanisms of cancer cachexia have provided a platform for the development of new targeted therapies. Although recent outcomes of early-phase trials have showed that several drugs presented an ideal curative effect, monotherapy cannot be entirely satisfactory in the treatment of cachexia-associated symptoms due to its complex and multifactorial pathogenesis. Therefore, the lack of definitive therapeutic strategies for cancer cachexia emphasizes the need to develop a better understanding of the underlying mechanisms. Increasing evidences show that the progression of cachexia is associated with metabolic alternations, which mainly include excessive energy expenditure, increased proteolysis and mitochondrial dysfunction. In this review, we provided an overview of the key mechanisms of cancer cachexia, with a major focus on muscle atrophy, adipose tissue wasting, anorexia and fatigue and updated the latest progress of pharmacological management of cancer cachexia, thereby further advancing the interventions that can counteract cancer cachexia.


Asunto(s)
Anorexia/tratamiento farmacológico , Antineoplásicos/uso terapéutico , Caquexia/tratamiento farmacológico , Fatiga/tratamiento farmacológico , Atrofia Muscular/tratamiento farmacológico , Neoplasias/tratamiento farmacológico , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/metabolismo , Tejido Adiposo/patología , Anorexia/complicaciones , Anorexia/metabolismo , Anorexia/mortalidad , Antiinflamatorios/uso terapéutico , Estimulantes del Apetito/uso terapéutico , Caquexia/complicaciones , Caquexia/metabolismo , Caquexia/mortalidad , Fatiga/complicaciones , Fatiga/metabolismo , Fatiga/mortalidad , Humanos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Mitocondrias/patología , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Atrofia Muscular/complicaciones , Atrofia Muscular/metabolismo , Atrofia Muscular/mortalidad , Neoplasias/complicaciones , Neoplasias/metabolismo , Neoplasias/mortalidad , Calidad de Vida , Análisis de Supervivencia , Congéneres de la Testosterona/uso terapéutico , Pérdida de Peso/efectos de los fármacos
8.
J Cell Physiol ; 236(5): 3445-3465, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33200401

RESUMEN

Tumor-associated macrophages (TAMs) in solid tumors exert protumor activities by releasing cytokines or growth factors into the tumor microenvironment. Increasing studies have also shown that TAMs play a key role in tumor progression, such as tumor angiogenesis, immunosuppression, cell proliferation, migration, invasion, and metastasis. A large body of evidence shows that the abundance of TAMs in solid tumors is correlated with poor disease prognosis and resistance to therapies. Therefore, targeting TAMs in solid tumors is considered to be a promising immunotherapeutic strategy. At present, the therapeutic strategies of targeting macrophages mainly include limiting monocyte recruitment, depletion strategies, promoting macrophage phagocytic activity, and induction of macrophage reprogramming. Additionally, targeting TAMs in combination with conventional therapies has been demonstrated to be a promising therapeutic strategy in solid tumors. In the present review, we summarized various TAMs-targeting therapeutic strategies for treating solid tumors. This review also discusses the challenges for targeting TAMs as tumor treatments, the obstacles in clinical trials, and the perspective for the future development of TAMs-targeting therapies for various cancers.


Asunto(s)
Neoplasias/patología , Neoplasias/terapia , Microambiente Tumoral/inmunología , Macrófagos Asociados a Tumores/inmunología , Animales , Proliferación Celular/fisiología , Humanos , Macrófagos/metabolismo , Neoplasias/inmunología , Neovascularización Patológica/patología
9.
Biochem Biophys Res Commun ; 558: 216-223, 2021 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-33008584

RESUMEN

BET inhibitor (BETi) has potential therapeutic effects on human cancer especially in breast cancer. However, the detailed mechanisms remain unclear. Herein, we found that BETi JQ1 and I-BET-151 (I-BET) activated ATF2 through JNK1/2 pathway in breast cancer cells MDA-MB-231 (MB-231). In addition, overexpression of ATF2 blocked the reduction of cell viability induced by JQ1 or I-BET in breast cancer MB-231 and BT-549 cells, cervical cancer HeLa cells and lung cancer A549 cells. The induction of cell death by BETi was also attenuated by ATF2 in MB-231 and BT-549 cells. By contrast, depletion of ATF2 increased cancer cell sensitivity to BETi. In MB-231 cells xenograft model, ATF2 significantly inhibited the anti-tumor effects of JQ1. By detection of the oxidized form gluthione, malondialdehyde and lipid ROS, we showed that overexpression of ATF2 inhibited ferroptosis induced by BETi, whereas depletion of ATF2 promoted ferroptosis by BETi. Furthermore, the underlying mechanisms of ATF2-reduced ferroptosis were investigated. Overexpressed and depleted ATF2 were found to significantly upregulate and downregulate NRF2 protein and mRNA expression, respectively. The significantly positive correlations between NRF2 and ATF2 gene expression were found in breast, lung and cervical cancer tissues from TCGA database. In NRF2-depleted MB-231 cells, ATF2 failed to attenuate JQ1-stimulated ferroptosis. All these results suggested that ATF2 inhibited BETi-induced ferroptosis by increasing NRF2 expression. Altogether, our findings illustrated ATF2 suppressed ani-tumor effects of BETi in a negative feedback manner by attenuating ferroptosis. BETi combined with ATF2 or NRF2 inhibitor might be a novel strategy for treatment of human cancer.


Asunto(s)
Factor de Transcripción Activador 2/metabolismo , Antineoplásicos/farmacología , Ferroptosis/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Proteínas/antagonistas & inhibidores , Células A549 , Factor de Transcripción Activador 2/deficiencia , Factor de Transcripción Activador 2/genética , Animales , Azepinas/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Resistencia a Antineoplásicos/efectos de los fármacos , Retroalimentación Fisiológica/efectos de los fármacos , Femenino , Células HeLa , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Ratones Desnudos , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Neoplasias/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Triazoles/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Apoptosis ; 25(3-4): 205-216, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31938895

RESUMEN

Emerging evidence has indicated that microRNAs are involved in multiple processes of cancer development. Previous studies have demonstrated that microRNA-499a (miR-499a) plays both oncogenic and tumor suppressive roles in several types of malignancies, and genetic variants in miR-499a are associated with the risk of cervical cancer. However, the biological roles of miR-499a in cervical cancer have not been investigated. Quantitative real-time PCR was used to assess miR-499a expression in cervical cancer cells. Mimics or inhibitor of miR-499a was transfected into cervical cancer cells to upregulate or downregulate miR-499a expression. The effects of miR-499a expression change on cervical cancer cells proliferation, colony formation, tumorigenesis, chemosensitivity, transwell migration and invasion were assessed. The potential targets of miR-499a were predicted using online database tools and validated using real-time PCR, Western blot and luciferase reporter experiments. miR-499a was significantly upregulated in cervical cancer cells. Moreover, overexpression of miR-499a significantly enhanced the proliferation, cell cycle progression, colony formation, apoptosis resistance, migration and invasion of cervical cancer cells, while inhibiting miR-499a showed the opposite effects. Further exploration demonstrated that Sex-determining region Y box 6 was the direct target of miR-499a. miR-499a-induced SOX6 downregulation mediated the oncogenic effects of miR-499a in cervical cancer. Inhibiting miR-499a could enhance the anticancer effects of cisplatin in the xenograft mouse model of cervical cancer. Our findings for the first time suggest that miRNA-499a may play an important role in the development of cervical cancer and could serve as a potential therapeutic target.


Asunto(s)
Resistencia a Antineoplásicos , MicroARNs/metabolismo , Factores de Transcripción SOXD/genética , Neoplasias del Cuello Uterino/patología , Animales , Apoptosis , Ciclo Celular , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Cisplatino/farmacología , Cisplatino/uso terapéutico , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , MicroARNs/antagonistas & inhibidores , MicroARNs/genética , Factores de Transcripción SOXD/metabolismo , Neoplasias del Cuello Uterino/genética , Neoplasias del Cuello Uterino/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Biochem Biophys Res Commun ; 533(4): 1122-1128, 2020 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-33036752

RESUMEN

Skeletal muscle and white adipose tissue are important organs of glucose-lipid metabolism. However, excessive lipolysis and free fatty acids (FFA) release in adipocytes elevate plasma FFA, leading to insulin resistance in skeletal muscle. Here, we investigated effects of insulin-resistant adipocytes on skeletal muscle in vitro by simulating body environment using a transwell coculture method. Insulin-resistant 3T3-L1 adipocytes increased lipolysis and FFA release, which reduced insulin sensitivity in the cocultured C2C12 myotubes. Rosiglitazone (RSG) decreased excessive lipolysis by reducing expression of adipose triglyceride lipase (ATGL) and activity of hormone-sensitive lipase (HSL), which led to decrease of FFA release from insulin-resistant 3T3-L1 adipocytes. Meanwhile, insulin resistance in C2C12 myotubes cocultured with insulin-resistant 3T3-L1 adipocytes was ameliorated after RSG treatment. Taken together, our present study provided direct evidence to better understand insulin resistance between skeletal muscle and adipose tissue in type 2 diabetes.


Asunto(s)
Adipocitos/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Ácidos Grasos no Esterificados/metabolismo , Resistencia a la Insulina/fisiología , Metabolismo de los Lípidos/efectos de los fármacos , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , Células 3T3-L1 , Adipocitos/efectos de los fármacos , Animales , Asialoglicoproteínas/genética , Asialoglicoproteínas/metabolismo , Comunicación Celular/fisiología , Técnicas de Cocultivo , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Ácidos Grasos no Esterificados/sangre , Hipoglucemiantes/farmacología , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Lipasa/genética , Lipasa/metabolismo , Lipólisis/efectos de los fármacos , Lipólisis/fisiología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Fibras Musculares Esqueléticas/efectos de los fármacos , Músculo Esquelético/efectos de los fármacos , Rosiglitazona/farmacología , Esterol Esterasa/genética , Esterol Esterasa/metabolismo
12.
Biochem Biophys Res Commun ; 530(2): 402-409, 2020 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-32536370

RESUMEN

BET inhibitors (BETi) exert an excellent anti-cancer activity in breast cancer. However, the identification of new potential targets to enhance breast cancer sensitivity to BETi is still an enormous challenge. Both NR5A2 and NCOA3 are frequently involved in cancer cells resistance to chemotherapy, also associated with poor prognosis in breast cancer. However, the functions of NR5A2 and NCOA3 in BETi resistance remains unknown. In this study, we found that BETi JQ1 and I-BET151 exhibited anti-cancer effects in breast cancer by inducing ferroptosis. NCOA3 as a coactivator synergized with NR5A2 to prevent BETi-induced ferroptosis. Mechanistically, we identified NR5A2 synergized with NCOA3 to increase expression of NRF2, a transcription factor that controls the expression of many antioxidant genes. Moreover, inhibition of NR5A2 or NCOA3 using small molecule inhibitors enhanced anti-cancer effects of BETi against breast cancer in vivo and in vitro. Altogether, our findings illustrated NR5A2 synergized with NCOA3 to confer breast cancer cells resistance to BETi by induction of NRF2. Inhibition of NR5A2/NCOA3 combined with BETi might be a novel strategy for treatment of breast cancer.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Ferroptosis/efectos de los fármacos , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Factor 2 Relacionado con NF-E2/genética , Coactivador 3 de Receptor Nuclear/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Resistencia a Antineoplásicos/efectos de los fármacos , Femenino , Humanos , Ratones , Ratones Desnudos , Factor 2 Relacionado con NF-E2/metabolismo , Coactivador 3 de Receptor Nuclear/antagonistas & inhibidores , Receptores Citoplasmáticos y Nucleares/antagonistas & inhibidores , Regulación hacia Arriba/efectos de los fármacos
13.
J Biomed Sci ; 26(1): 78, 2019 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-31629410

RESUMEN

In many solid tumor types, tumor-associated macrophages (TAMs) are important components of the tumor microenvironment (TME). Moreover, TAMs infiltration is strongly associated with poor survival in solid tumor patients. In this review, we describe the origins of TAMs and their polarization state dictated by the TME. We also specifically focus on the role of TAMs in promoting tumor growth, enhancing cancer cells resistance to chemotherapy and radiotherapy, promoting tumor angiogenesis, inducing tumor migration and invasion and metastasis, activating immunosuppression. In addition, we discuss TAMs can be used as therapeutic targets of solid tumor in clinics. The therapeutic strategies include clearing macrophages and inhibiting the activation of TAMs, promoting macrophage phagocytic activity, limiting monocyte recruitment and other targeted TAMs therapies.


Asunto(s)
Progresión de la Enfermedad , Macrófagos/microbiología , Neoplasias/inmunología , Animales , Humanos , Ratones , Neoplasias/etiología , Neoplasias/fisiopatología , Neovascularización Patológica , Microambiente Tumoral
14.
Apoptosis ; 22(11): 1321-1335, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28936716

RESUMEN

Reactive oxygen species (ROS), a group of ions and molecules, include hydroxyl radicals (·OH), alkoxyl radicals, superoxide anion (O2·-), singlet oxygen (1O2) and hydrogen peroxide (H2O2). Hydroxyl radicals and alkoxyl radicals are extremely and highly reactive species respectively. Endogenous ROS are mainly formed in mitochondrial respiratory chain. Low levels of ROS play important roles in regulating biological functions in mammalian cells. However, excess production of ROS can induce cell death by oxidative damaging effects to intracellular biomacromolecules. Cancer cell death types induced by ROS include apoptotic, autophagic, ferroptotic and necrotic cell death. Since abnormal metabolism in cancer cells, they have higher ROS content compared to normal cells. The higher endogenous ROS levels in cancer cells endow them more susceptible to the ROS-induction treatment. Indeed, some anticancer drugs currently used in clinic, such as molecular targeted drugs and chemotherapeutic agents, effectively kill cancer cells by inducing ROS generation. In addition, photodynamic therapy (PDT) is mainly based on induction of ROS burst to kill cancer cells. The mechanism of cell death induced by radiotherapy using ionizing radiation also refers to ROS production. Moreover, ROS play an important role in tumor immune therapy. Altogether, combining above traditional treatments with ROS-induced agents will be considered as a promising strategy in cancer therapy. In this review, we focus on our current understanding of the anticancer effects of ROS.


Asunto(s)
Proteínas del Complejo de Cadena de Transporte de Electrón/genética , Regulación Neoplásica de la Expresión Génica , Terapia Molecular Dirigida/métodos , Neoplasias/tratamiento farmacológico , Neoplasias/radioterapia , Especies Reactivas de Oxígeno/agonistas , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Mitocondrias/efectos de la radiación , Necrosis/inducido químicamente , Necrosis/genética , Necrosis/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Fotoquimioterapia , Fármacos Fotosensibilizantes/uso terapéutico , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Radiación Ionizante , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal
15.
Biochem Biophys Res Commun ; 478(1): 227-233, 2016 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-27425252

RESUMEN

SRC-3 is widely expressed in multiple tumor types and involved in cancer cell proliferation and apoptosis. Histone deacetylase (HDAC) inhibitors are promising antitumor drugs. However, the poor efficacy of HDAC inhibitors in solid tumors has restricted its further clinical application. Here, we reported the novel finding that depletion of SRC-3 enhanced sensitivity of breast and lung cancer cells to HDAC inhibitors (SAHA and romidepsin). In contrast, overexpression of SRC-3 decreased SAHA-induced cancer cell apoptosis. Furthermore, we found that SRC-3 inhibitor bufalin increased cancer cell apoptosis induced by HDAC inhibitors. The combination of bufalin and SAHA was particular efficient in attenuating AKT activation and reducing Bcl-2 levels. Taken together, these accumulating data might guide development of new breast and lung cancer therapies.


Asunto(s)
Antibióticos Antineoplásicos/administración & dosificación , Inhibidores de Histona Desacetilasas/administración & dosificación , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/metabolismo , Coactivador 3 de Receptor Nuclear/genética , Coactivador 3 de Receptor Nuclear/metabolismo , Células A549 , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Depsipéptidos/administración & dosificación , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Silenciador del Gen , Humanos , Neoplasias Experimentales/patología , Coactivador 3 de Receptor Nuclear/antagonistas & inhibidores
16.
Cell Physiol Biochem ; 35(6): 2255-71, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25895606

RESUMEN

BACKGROUND/AIM: Treatment of human non-small-cell lung cancer (NSCLC) often involves uses of multiple therapeutic strategies with different mechanisms of action. Here we found that resveratrol (RV) enhanced the anti-tumor effects of epidermal growth factor receptor (EGFR) inhibitor erlotinib in NSCLC cells. METHODS: Cell viability was measured by MTT assay and clonogenicity assay. Western blot was applied to assess the protein expression levels of target genes. Cell apoptosis was monitored by AnnexinV-FITC assay and sub-G1 population assay. Intracellular ROS were measured by flow cytometric analysis. Cell caspase activities were carried out by fluorometric assays. RESULTS: Exposure of H460, A549, PC-9 and H1975 cells to minimal or non-toxic concentrations of RV and erlotinib synergistically reduced cell viability, colony formation and induced cell apoptosis. Furthermore, RV synergistically enhanced erlotinib-induced apoptosis was involved in ROS production. Additionally, co-treatment with RV and erlotinib repressed the expressions of anti-apoptosis proteins, such as survivin and Mcl-1, whereas promoted p53 and PUMA expression and caspase 3 activity. Moreover, the combination was also more effective at inhibiting the AKT/mTOR/S6 kinase pathway. Subsequently, small interfering RNA (siRNA) depletion of PUMA and overexpression of survivin significantly attenuated NSCLC cells apoptosis induced by the combination of the two drugs. CONCLUSION: Our findings suggested that RV synergistically enhanced the anti-tumor effects of erlotinib in NSCLC cells were involved in decrease of survivin expression and induction of PUMA expression. In conclusion, based on the observations from our study, we indicated that the combined administration of these two drugs might be considered as a novel therapeutic regimen for treating NSCLC.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Apoptosis/efectos de los fármacos , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Clorhidrato de Erlotinib/farmacología , Proteínas Inhibidoras de la Apoptosis/metabolismo , Neoplasias Pulmonares/tratamiento farmacológico , Proteínas Proto-Oncogénicas/metabolismo , Estilbenos/farmacología , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Caspasa 3/metabolismo , Línea Celular Tumoral , Regulación hacia Abajo/efectos de los fármacos , Sinergismo Farmacológico , Humanos , Neoplasias Pulmonares/metabolismo , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Resveratrol , Proteínas Quinasas S6 Ribosómicas/metabolismo , Transducción de Señal/efectos de los fármacos , Survivin , Serina-Treonina Quinasas TOR/metabolismo , Proteína p53 Supresora de Tumor , Regulación hacia Arriba/efectos de los fármacos
17.
Acta Pharm Sin B ; 13(6): 2645-2662, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37425043

RESUMEN

Induction of cancer cell ferroptosis has been proposed as a potential treatment in several cancer types. Tumor-associated macrophages (TAMs) play a key role in promoting tumor malignant progression and therapy resistance. However, the roles and mechanisms of TAMs in regulating tumor ferroptosis is still unexplored and remains enigmatic. This study shows ferroptosis inducers has shown therapeutic outcomes in cervical cancer in vitro and in vivo. TAMs have been found to suppress cervical cancer cells ferroptosis. Mechanistically, macrophage-derived miRNA-660-5p packaged into exosomes are transported into cancer cells. In cancer cells, miRNA-660-5p attenuates ALOX15 expression to inhibit ferroptosis. Moreover, the upregulation of miRNA-660-5p in macrophages depends on autocrine IL4/IL13-activated STAT6 pathway. Importantly, in clinical cervical cancer cases, ALOX15 is negatively associated with macrophages infiltration, which also raises the possibility that macrophages reduce ALOX15 levels in cervical cancer. Moreover, both univariate and multivariate Cox analyses show ALOX15 expression is independent prognostic factor and positively associated with good prognosis in cervical cancer. Altogether, this study reveals the potential utility of targeting TAMs in ferroptosis-based treatment and ALOX15 as prognosis indicators for cervical cancer.

18.
Biomed Pharmacother ; 161: 114567, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36963362

RESUMEN

Immune checkpoint blockade (ICB) therapy targeting the programmed death 1/programmed death-ligand 1 (PD-1/PD-L1) axis has achieved considerable success in treating a wide range of cancers. However, most patients with pancreatic cancer remain resistant to ICB. Moreover, there is a lack of optimal biomarkers for the prediction of response to this therapy. Palmitoylation is mediated by a family of 23 S-acyltransferases, termed zinc finger Asp-His-His-Cys-type palmitoyltransferases (ZDHHC), which precisely control various cancer-related protein functions and represent promising drug targets for cancer therapy. Here, we revealed that tumor cell-intrinsic ZDHHC9 was overexpressed in pancreatic cancer tissues and associated with impaired anti-tumor immunity. In syngeneic pancreatic tumor models, the knockdown of ZDHHC9 expression suppressed tumor progression and prolonged survival time of mice by modifying the immunosuppressive ('cold') to proinflammatory ('hot') tumor microenvironment. Furthermore, ZDHHC9 deficiency sensitized anti-PD-L1 immunotherapy mainly in a CD8+ T cell dependent manner. Lastly, we employed the ZDHHC9-siRNA nanoparticle system to efficiently silence ZDHHC9 in pancreatic tumors. Collectively, our findings indicate that ZDHHC9 overexpression in pancreatic tumors is a mechanism involved in the inhibition of host anti-tumor immunity and highlight the importance of inactivating ZDHHC9 as an effective immunotherapeutic strategy and booster for anti-PD-L1 therapy against pancreatic cancer.


Asunto(s)
Neoplasias Pancreáticas , Microambiente Tumoral , Animales , Ratones , Aciltransferasas/genética , Inmunoterapia , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas
19.
J Transl Med ; 9: 74, 2011 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-21600017

RESUMEN

BACKGROUND: Aurora kinase ensures accurate chromosome segregation during cell cycle, maintaining genetic integrity in cell division. VX-680, a small-molecule Aurora kinase inhibitor, interferes with mitotic entry and formation of bipolar spindles. Here, we evaluated VX-680 as a potential agent for treatment of all-trans retinoid acid (ATRA)-resistant acute promyelocytic leukemia (APL) in vitro. METHODS: CD11b expression was utilized to assess cell differentiation by flow cytometry. Immunofluorescence staining was conducted to analyze formation of cell monopolar spindle. Cell proliferation was evaluated by MTT assay. Sub-G1 population and Annexin V/PI staining were used to measure cell apoptosis. Hoechst 33342 staining was applied for identifying morphological changes in nucleus of apoptotic cell. Aurora-A (Aur-A) activation and the signaling pathways involved in apoptosis were detected by Western blot. JC-1 probe was employed to measure mitochondrial depolarization. RESULTS: VX-680 inhibited Aur-A by reducing autophosphorylation at the activation site, Thr288, accompanied by producing monopolar mitotic spindles in APL cell line NB4-R2 that was resistant to ATRA. In addition, we found that VX-680 inhibited cell proliferation as assessed by MTT assay. Flow cytometry showed that VX-680 led to apoptotic cell death in both dose- and time-dependent manners by either Sub-G1 or Annexin V/PI analysis. Hoechst 33342 staining represented typical apoptotic cells with nuclear fragmentation in VX-680 treated cells. Importantly, VX-680 inhibition of Aurora kinase suppressed Akt-1 activation and induced mitochondrial depolarization, which eventually resulted in apoptosis by activation of caspase pathway, as indicated by increasing proteolytic cleavage of procaspase-3 and poly ADP ribose polymerase (PARP) in NB4-R2 cells. CONCLUSIONS: Our study suggested potential clinical use of mitotic Aurora kinase inhibitor in targeting ATRA-resistant leukemic cells.


Asunto(s)
Apoptosis/efectos de los fármacos , Leucemia Promielocítica Aguda/patología , Mitosis/efectos de los fármacos , Piperazinas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/metabolismo , Tretinoina/farmacología , Anexina A5/metabolismo , Aurora Quinasas , Caspasas/metabolismo , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Núcleo Celular/efectos de los fármacos , Núcleo Celular/patología , Proliferación Celular/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Humanos , Leucemia Promielocítica Aguda/enzimología , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Propidio/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacología , Huso Acromático/efectos de los fármacos , Huso Acromático/metabolismo
20.
Aging (Albany NY) ; 13(7): 10015-10033, 2021 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-33795530

RESUMEN

Ameliorating hyperglycemia and insulin resistance are major therapeutic strategies for type 2 diabetes. Previous studies have indicated that photobiomodulation therapy (PBMT) attenuates metabolic abnormalities in insulin-resistant adipose cells and tissues. However, it remains unclear whether PBMT ameliorates glucose metabolism in skeletal muscle in type 2 diabetes models. Here we showed that PBMT reduced blood glucose and insulin resistance, and reversed metabolic abnormalities in skeletal muscle in two diabetic mouse models. PBMT accelerated adenosine triphosphate (ATP) and reactive oxygen species (ROS) generation by elevating cytochrome c oxidase (CcO) activity. ROS-induced activation of phosphatase and tensin homolog (PTEN)/ protein kinase B (AKT) signaling after PBMT promoted glucose transporter GLUT4 translocation and glycogen synthase (GS) activation, accelerating glucose uptake and glycogen synthesis in skeletal muscle. CcO subunit III deficiency, ROS elimination, and AKT inhibition suppressed the PBMT effects of glucose metabolism in skeletal muscle. This study indicated amelioration of glucose metabolism after PBMT in diabetic mouse models and revealed the metabolic regulatory effects and mechanisms of PBMT on skeletal muscle.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Complejo IV de Transporte de Electrones/metabolismo , Hiperglucemia/terapia , Resistencia a la Insulina/fisiología , Terapia por Luz de Baja Intensidad , Músculo Esquelético/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Línea Celular , Glucosa/metabolismo , Hiperglucemia/metabolismo , Ratones , Fosfohidrolasa PTEN/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/fisiología , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA